MECP2

MECP2
  • 文章类型: Journal Article
    表观遗传学是研究基因组和基因表达模式的可遗传变化,这些变化不是由DNA序列的直接变化引起的。这些变化的例子包括对DNA结合的组蛋白的翻译后修饰,DNA甲基化,和重建核架构。总的来说,表观遗传变化提供了一层调控,影响基因的转录活性,同时保持DNA序列不变。已经在先天性心脏病(CHD)患者中发现了影响负责修饰或感知表观遗传标记的酶的序列变异或突变。和表观遗传复合物的小分子抑制剂已显示出有望作为成人心脏病的疗法。此外,具有编码表观遗传酶的基因突变或缺失的转基因小鼠概括了人类心脏病的各个方面。一起来看,这些研究结果表明,表观遗传学领域的发展将为我们理解先天性和成人心脏病提供新的治疗机会.
    Epigenetics is the study of heritable changes to the genome and gene expression patterns that are not caused by direct changes to the DNA sequence. Examples of these changes include posttranslational modifications to DNA-bound histone proteins, DNA methylation, and remodeling of nuclear architecture. Collectively, epigenetic changes provide a layer of regulation that affects transcriptional activity of genes while leaving DNA sequences unaltered. Sequence variants or mutations affecting enzymes responsible for modifying or sensing epigenetic marks have been identified in patients with congenital heart disease (CHD), and small-molecule inhibitors of epigenetic complexes have shown promise as therapies for adult heart diseases. Additionally, transgenic mice harboring mutations or deletions of genes encoding epigenetic enzymes recapitulate aspects of human cardiac disease. Taken together, these findings suggest that the evolving field of epigenetics will inform our understanding of congenital and adult cardiac disease and offer new therapeutic opportunities.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    神经发育障碍PittHopkins综合征(PTHS)引起与Rett综合征(RTT)患者相似的临床症状。然而,RTT由MECP2突变引起,而TCF4基因中的突变导致PTHS。这两种疾病背后的机械共性是未知的,但是他们的共同症状表明,趋同通路水平的破坏可能存在。我们将患者皮肤来源的成纤维细胞重编程为诱导的神经元祖细胞。有趣的是,我们发现,与健康对照组相比,PTHS患者iNPC中MeCP2水平降低,iNPC和i星形胶质细胞均以突变特异性方式显示功能和分化缺陷.当Tcf4+/-小鼠与过表达MeCP2的小鼠遗传杂交时,分子和表型缺陷显着改善,MeCP2在PTHS病理中的强调和重要作用。重要的是,出生后脑室内基因替代疗法,使用表达9型腺相关病毒载体(AAV9)的MeCP2(AAV9。P546.MeCP2)显着改善了iNPC和i星形胶质细胞的功能,并有效改善了Tcf4/-小鼠的组织学和行为缺陷。合并,我们的数据提示MeCP2在PTHS病理和常见通路中的作用之前未知,可能在多种神经发育障碍中受到影响.我们的工作突出了PTHS的潜在新治疗靶点,包括上调MeCP2表达或其下游靶标,潜在的,基于MeCP2的基因治疗。
    The neurodevelopmental disorder Pitt Hopkins syndrome (PTHS) causes clinical symptoms similar to Rett syndrome (RTT) patients. However, RTT is caused by MECP2 mutations whereas mutations in the TCF4 gene lead to PTHS. The mechanistic commonalities underling these two disorders are unknown, but their shared symptomology suggest that convergent pathway-level disruption likely exists. We reprogrammed patient skin derived fibroblasts into induced neuronal progenitor cells. Interestingly, we discovered that MeCP2 levels were decreased in PTHS patient iNPCs relative to healthy controls and that both iNPCs and iAstrocytes displayed defects in function and differentiation in a mutation-specific manner. When Tcf4+/- mice were genetically crossed with mice overexpressing MeCP2, molecular and phenotypic defects were significantly ameliorated, underlining and important role of MeCP2 in PTHS pathology. Importantly, post-natal intracerebroventricular gene replacement therapy with adeno-associated viral vector serotype 9 (AAV9)-expressing MeCP2 (AAV9.P546.MeCP2) significantly improved iNPC and iAstrocyte function and effectively ameliorated histological and behavioral defects in Tcf4+/- mice. Combined, our data suggest a previously unknown role of MeCP2 in PTHS pathology and common pathways that might be affected in multiple neurodevelopmental disorders. Our work highlights potential novel therapeutic targets for PTHS, including upregulation of MeCP2 expression or its downstream targets or, potentially, MeCP2-based gene therapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    Nodding综合征是一种与神经炎症和tau蛋白病相关的癫痫性脑病。最初是小儿脑部疾病,与甲基-CpG结合蛋白2(MECP2)重复综合征有一些临床重叠,影响了某些贫困的东非社区,同时发生了当地的内战和国内流离失所,迫使人们依赖受污染的食物和水的条件。某些生物毒素(淡水蓝藻毒素加/减霉菌毒素)与神经炎症在Nodding综合征中的潜在作用,兴奋毒性,Tau病法,和MECP2失调特性,在这里是第一次考虑。
    Nodding syndrome is an epileptic encephalopathy associated with neuroinflammation and tauopathy. This initially pediatric brain disease, which has some clinical overlap with Methyl-CpG-binding protein 2 (MECP2) Duplication Syndrome, has impacted certain impoverished East African communities coincident with local civil conflict and internal displacement, conditions that forced dependence on contaminated food and water. A potential role in Nodding syndrome for certain biotoxins (freshwater cyanotoxins plus/minus mycotoxins) with neuroinflammatory, excitotoxic, tauopathic, and MECP2-dysregulating properties, is considered here for the first time.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    目的:甲基CpG结合蛋白2(MECP2)重复综合征是一种罕见的X连锁基因组疾病,主要影响男性,通常表现为癫痫和自闭症谱系障碍(ASD)合并症。转基因系MeCP2Tg1用于模拟MECP2重复综合征,并显示自闭症-癫痫并存。先前的工作表明,兴奋性/抑制性(E/I)失衡是癫痫和ASD的潜在共同机制。投射神经元和小白蛋白(PV)中间神经元占海马中E/I平衡的大部分。因此,我们探讨了MeCP2Tg1小鼠海马中投射和PV+神经元的结构变化是如何发生的,以及这些形态学变化是否有助于癫痫易感性.
    方法:我们使用设计药物小鼠模型专门激活的中间神经元Designer受体来抑制海马中的抑制性神经元,以验证MeCP2Tg1的癫痫易感性(FVB,一种近交系,称为对Friend白血病病毒敏感)小鼠。记录脑电图以定义癫痫发作。我们在MeCP2Tg1(FVB):CaMKIIα-Cre(C57BL/6)小鼠或MeCP2Tg1:PV-Cre(C57BL/6)小鼠及其同窝对照中进行了病毒的眼眶后注射,以特异性标记投射和PV神经元进行结构分析。
    结果:MeCP2Tg1小鼠癫痫易感性增加。MeCP2Tg1小鼠海马中PV神经元数量减少,树突复杂性降低。与野生型小鼠相比,MeCP2Tg1小鼠的树突复杂性增加,MeCP2Tg1小鼠齿状回总树突棘密度也增加。MeCP2Tg1小鼠的CA1中总树突棘密度增加。
    结论:MeCP2的过表达可能会破坏关键的信号通路,导致PV中间神经元的树突复杂性降低,投射神经元的树突脊柱密度增加。与MeCP2相关的兴奋性和抑制性神经元结构的这种相互调节暗示了其作为癫痫发展中的潜在靶标的重要性,并为自闭症和癫痫的共同发生提供了新的视角。
    OBJECTIVE: Methyl CpG-binding protein 2 (MECP2) duplication syndrome is a rare X-linked genomic disorder affecting predominantly males, which is usually manifested as epilepsy and autism spectrum disorder (ASD) comorbidity. The transgenic line MeCP2Tg1 was used for mimicking MECP2 duplication syndrome and showed autism-epilepsy co-occurrence. Previous works suggested that the excitatory/inhibitory (E/I) imbalance is a potential common mechanism for both epilepsy and ASD. The projection neurons and parvalbumin (PV) interneurons account for the majority of E/I balance in the hippocampus. Therefore, we explored how structural changes of projection and PV+ neurons occur in the hippocampus of MeCP2Tg1 mice and whether these morphological changes contribute to epilepsy susceptibility.
    METHODS: We used the interneuron Designer receptors exclusively activated by designer drugs mouse model to inhibit inhibitory neurons in the hippocampus to verify the epilepsy susceptibility of MeCP2Tg1 (FVB, an inbred strain named as sensitivity to Friend leukemia virus) mice. Electroencephalograms were recorded for the definition of seizure. We performed retro-orbital injection of virus in MeCP2Tg1 (FVB):CaMKIIα-Cre (C57BL/6) mice or MeCP2Tg1:PV-Cre (C57BL/6) mice and their littermate controls to specifically label projection and PV+ neurons for structural analysis.
    RESULTS: Epilepsy susceptibility was increased in MeCP2Tg1 mice. There was a reduced number of PV neurons and reduced dendritic complexity in the hippocampus of MeCP2Tg1 mice. The dendritic complexity in MeCP2Tg1 mice was increased compared to wild-type mice, and total dendritic spine density in dentate gyrus of MeCP2Tg1 mice was also increased. Total dendritic spine density was increased in CA1 of MeCP2Tg1 mice.
    CONCLUSIONS: Overexpression of MeCP2 may disrupt crucial signaling pathways, resulting in decreased dendritic complexity of PV interneurons and increased dendritic spine density of projection neurons. This reciprocal modulation of excitatory and inhibitory neuronal structures associated with MeCP2 implies its significance as a potential target in the development of epilepsy and offers a novel perspective on the co-occurrence of autism and epilepsy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    迄今为止,Rett综合征(RTT),主要由X连锁MECP2基因突变引起的遗传性疾病,越来越被认为是一种广谱病理学,而不仅仅是神经发育疾病,由于大量的外周共病和受损的代谢途径,影响患者。改变的分子过程包括受损的线粒体功能,受干扰的氧化还原稳态,慢性亚临床炎症和不适当的胆固醇代谢。10年前首次定义了持续性亚临床炎症,作为以前无法识别的RTT功能,在病理进展和表型严重程度的调节中起作用。鉴于此,本工作旨在回顾目前有关RTT中慢性炎症状态和免疫/炎症功能改变的知识,以及调查这种疾病背后的新机制,特别关注关于炎症小体系统的最新发现,自身免疫反应和肠道微生物和真菌群。在这些基础上,虽然还需要进一步的研究,能够重新建立足够免疫/炎症反应的未来治疗策略可能是RTT患者的潜在方法.
    To date, Rett syndrome (RTT), a genetic disorder mainly caused by mutations in the X-linked MECP2 gene, is increasingly considered a broad-spectrum pathology, instead of just a neurodevelopmental disease, due to the multitude of peripheral co-morbidities and the compromised metabolic pathways, affecting the patients. The altered molecular processes include an impaired mitochondrial function, a perturbed redox homeostasis, a chronic subclinical inflammation and an improper cholesterol metabolism. The persistent subclinical inflammatory condition was first defined ten years ago, as a previously unrecognized feature of RTT, playing a role in the pathology progress and modulation of phenotypical severity. In light of this, the present work aims at reviewing the current knowledge on the chronic inflammatory status and the altered immune/inflammatory functions in RTT, as well as investigating the emerging mechanisms underlying this condition with a special focus on the latest findings about inflammasome system, autoimmunity responses and intestinal micro- and mycobiota. On these bases, although further research is needed, future therapeutic strategies able to re-establish an adequate immune/inflammatory response could represent potential approaches for RTT patients.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    Rett综合征(RTT)是由MECP2突变引起的神经发育障碍,它编码甲基CpG结合蛋白2,是许多基因的转录调节因子,包括脑源性神经营养因子(BDNF)。在Mecp2缺陷小鼠的多个脑区,BDNF水平较低,通过实验增加BDNF水平可以改善Mecp2突变小鼠的非典型表型。由于BDNF本身的血脑屏障通透性较低,我们测试了LM22A-4的效果,一种脑渗透剂,BDNF受体TrkB的小分子配体(由Ntrk2编码),对雌性Mecp2杂合(HET)小鼠的海马锥体神经元的树突棘密度和形态以及行为表型的影响。用LM22A-4对Mecp2HET小鼠进行为期4周的全身治疗,将MeCP2表达神经元的脊柱体积恢复到野生型(WT)水平,而缺乏MeCP2的神经元的脊柱体积仍与雌性WT小鼠的神经元相当。雌性Mecp2HET小鼠比WT小鼠更有攻击行为,通过4周的LM22A-4治疗,其水平降低至WT水平。这些数据为新疗法不仅对RTT而且对其他BDNF相关疾病的潜在有用性提供了额外的支持。
    Rett syndrome (RTT) is a neurodevelopmental disorder caused by mutations in MECP2, which encodes methyl-CpG-binding protein 2, a transcriptional regulator of many genes, including brain-derived neurotrophic factor (BDNF). BDNF levels are lower in multiple brain regions of Mecp2-deficient mice, and experimentally increasing BDNF levels improve atypical phenotypes in Mecp2 mutant mice. Due to the low blood-brain barrier permeability of BDNF itself, we tested the effects of LM22A-4, a brain-penetrant, small-molecule ligand of the BDNF receptor TrkB (encoded by Ntrk2), on dendritic spine density and form in hippocampal pyramidal neurons and on behavioral phenotypes in female Mecp2 heterozygous (HET) mice. A 4-week systemic treatment of Mecp2 HET mice with LM22A-4 restored spine volume in MeCP2-expressing neurons to wild-type (WT) levels, whereas spine volume in MeCP2-lacking neurons remained comparable to that in neurons from female WT mice. Female Mecp2 HET mice engaged in aggressive behaviors more than WT mice, the levels of which were reduced to WT levels by the 4-week LM22A-4 treatment. These data provide additional support to the potential usefulness of novel therapies not only for RTT but also to other BDNF-related disorders.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    结构变体(SV)的检测目前偏向于改变拷贝数的那些。倒位对遗传疾病的相对贡献尚不清楚。在这项研究中,我们分析了来自100,000基因组计划的33,924个罕见疾病家庭的基因组测序数据.从托管超过5亿个SV的数据库中,我们专注于351个基因,其中单倍体功能不全是已确认的疾病机制,并确定了47个超罕见重排,包括倒置(24bp至36.4Mb,20/47从头)。验证使用了许多正交方法,包括回顾性外显子组分析。RNA-seq数据支持六名参与者的各自诊断。表型混合在四个先证中很明显。诊断异常是一个共同的主题(一个人>50年),和特定基因的有针对性的分析已经进行了30%的这些个体,但没有发现。我们为基因内MSH2反演提供了欧洲创始人的正式确认。对于两个具有涉及MECP2突变热点的复杂SV的个体,使用长读数测序解决了模糊的SV结构,影响临床解释。在一个患有Kantaputra型中膜发育不良的家庭中发现了HOXD11-13的从头倒置。最后,一个复杂的易位干扰APC并涉及9个重排的节段,证实了3个家庭成员的临床诊断,并解决了一个患有单个息肉的兄弟姐妹的难题.总的来说,倒置在罕见疾病中起着很小但值得注意的作用,可能解释了大约1/750个家庭在不同临床队列中的病因。
    Detection of structural variants (SVs) is currently biased toward those that alter copy number. The relative contribution of inversions toward genetic disease is unclear. In this study, we analyzed genome sequencing data for 33,924 families with rare disease from the 100,000 Genomes Project. From a database hosting >500 million SVs, we focused on 351 genes where haploinsufficiency is a confirmed disease mechanism and identified 47 ultra-rare rearrangements that included an inversion (24 bp to 36.4 Mb, 20/47 de novo). Validation utilized a number of orthogonal approaches, including retrospective exome analysis. RNA-seq data supported the respective diagnoses for six participants. Phenotypic blending was apparent in four probands. Diagnostic odysseys were a common theme (>50 years for one individual), and targeted analysis for the specific gene had already been performed for 30% of these individuals but with no findings. We provide formal confirmation of a European founder origin for an intragenic MSH2 inversion. For two individuals with complex SVs involving the MECP2 mutational hotspot, ambiguous SV structures were resolved using long-read sequencing, influencing clinical interpretation. A de novo inversion of HOXD11-13 was uncovered in a family with Kantaputra-type mesomelic dysplasia. Lastly, a complex translocation disrupting APC and involving nine rearranged segments confirmed a clinical diagnosis for three family members and resolved a conundrum for a sibling with a single polyp. Overall, inversions play a small but notable role in rare disease, likely explaining the etiology in around 1/750 families across heterogeneous clinical cohorts.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Case Reports
    典型(或经典)Rett综合征(RTT)是一种X连锁神经发育障碍,其特征是一段时间的消退,部分或完全丧失有目的的手部动作,和后天的言语,步态受损,和刻板的手部动作。在超过95%的典型RTT中,在甲基-CPG结合蛋白2基因(MECP2)中发现了致病变体。这里,我们描述了一名临床诊断为典型RTT综合征的年轻女性,尽管进行了20年的调查和多轮MECP2基因测序,但她仍缺乏基因诊断.最近,使用下一代测序的额外基因检测已经完成,这揭示了BCL11A基因在MECP2的外显子4中的部分插入,导致MECP2中的小缺失,由于移码而可能导致MeCP2功能的破坏。这个案例展示了基因检测不断变化的局限性,以及随着技术的改进和更广泛的应用,不断追求诊断的重要性。
    Typical (or classic) Rett syndrome (RTT) is an X-linked neurodevelopmental disorder characterized by a period of regression, partial or complete loss of purposeful hand movements, and acquired speech, impaired gait, and stereotyped hand movements. In over 95% of typical RTT, a pathogenic variant is found in the methyl-CPG binding protein 2 gene (MECP2). Here, we describe a young woman with clinically diagnosed typical RTT syndrome who lacked a genetic diagnosis despite 20 years of investigation and multiple rounds of sequencing the MECP2 gene. Recently, additional genetic testing using next-generation sequencing was completed, which revealed a partial insertion of the BCL11A gene within exon 4 of MECP2, resulting in a small deletion in MECP2, causing likely disruption of MeCP2 function due to a frameshift. This case demonstrates the ever-changing limitations of genetic testing, as well as the importance of continual pursuit of a diagnosis as technologies improve and are more widely utilized.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    静止(G0)的维持和退出对于哺乳动物的组织稳态和再生至关重要。这里,我们表明,甲基-CpG结合蛋白2(Mecp2)的表达是细胞周期依赖性的,并且在培养的细胞和损伤诱导的肝再生小鼠模型中负调节静止退出。具体来说,随着Mecp2缺失加速,Mecp2的急性减少是有效的静止退出所必需的,而Mecp2的过表达延迟了静止退出,Mecp2条件性敲除后强制表达Mecp2可挽救细胞周期重新进入。E3连接酶Nedd4介导Mecp2的泛素化和降解,从而促进静止退出。一项全基因组研究揭示了Mecp2在通过转录激活代谢基因同时抑制增殖相关基因来防止静止退出中的双重作用。特别是两个核受体的破坏,Rara或Nr1h3,加速静止出口,模仿Mecp2耗竭表型。我们的研究揭示了Mecp2作为静止退出和组织再生的重要调节剂的先前未被认可的作用。
    Quiescence (G0) maintenance and exit are crucial for tissue homeostasis and regeneration in mammals. Here, we show that methyl-CpG binding protein 2 (Mecp2) expression is cell cycle-dependent and negatively regulates quiescence exit in cultured cells and in an injury-induced liver regeneration mouse model. Specifically, acute reduction of Mecp2 is required for efficient quiescence exit as deletion of Mecp2 accelerates, while overexpression of Mecp2 delays quiescence exit, and forced expression of Mecp2 after Mecp2 conditional knockout rescues cell cycle reentry. The E3 ligase Nedd4 mediates the ubiquitination and degradation of Mecp2, and thus facilitates quiescence exit. A genome-wide study uncovered the dual role of Mecp2 in preventing quiescence exit by transcriptionally activating metabolic genes while repressing proliferation-associated genes. Particularly disruption of two nuclear receptors, Rara or Nr1h3, accelerates quiescence exit, mimicking the Mecp2 depletion phenotype. Our studies unravel a previously unrecognized role for Mecp2 as an essential regulator of quiescence exit and tissue regeneration.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    MECP2重复综合征(MDS)是由至少MECP2和IRAK1基因的剂量增加引起的X连锁神经发育障碍,其特征是智力障碍(ID)。发育迟缓,低张力,癫痫和反复感染。它主要影响男性,女性可能受到影响或无症状携带者。Rett综合征(RTT)主要由MECP2中功能突变的丧失引发,是一种描述良好的综合征,表现为ID,癫痫,缺乏有目的的手使用和言语受损,在其他人中。由于实施了组学技术,已经报道了人类RTT样品中改变的生物途径,但这种分子特征尚未在MDS患者中进行。我们收集了17名MDS患者的皮肤成纤维细胞,10例MECP2重复携带者母亲和21例RTT患者,并进行了多组学(RNAseq和蛋白质组学)分析。这里,我们提供了详尽的描述,并比较了队列之间共同的和具体的失调的生物过程.我们还强调了基因TMOD2,SRGAP1,COPS2,CNPY2,IGF2BP1,MOB2,VASP,FZD7、ECSIT和KIF3B由于其在神经元功能中的暗示而作为生物标志物和治疗靶标候选物。定义RNA和蛋白质谱表明,我们的四个队列比它们共同的表型所预期的要少。
    MECP2 duplication syndrome (MDS) is an X-linked neurodevelopmental disorder caused by the gain of dose of at least the genes MECP2 and IRAK1 and is characterised by intellectual disability (ID), developmental delay, hypotonia, epilepsy and recurrent infections. It mainly affects males, and females can be affected or asymptomatic carriers. Rett syndrome (RTT) is mainly triggered by loss of function mutations in MECP2 and is a well described syndrome that presents ID, epilepsy, lack of purposeful hand use and impaired speech, among others. As a result of implementing omics technology, altered biological pathways in human RTT samples have been reported, but such molecular characterisation has not been performed in patients with MDS. We gathered human skin fibroblasts from 17 patients with MDS, 10 MECP2 duplication carrier mothers and 21 patients with RTT, and performed multi-omics (RNAseq and proteomics) analysis. Here, we provide a thorough description and compare the shared and specific dysregulated biological processes between the cohorts. We also highlight the genes TMOD2, SRGAP1, COPS2, CNPY2, IGF2BP1, MOB2, VASP, FZD7, ECSIT and KIF3B as biomarker and therapeutic target candidates due to their implication in neuronal functions. Defining the RNA and protein profiles has shown that our four cohorts are less alike than expected by their shared phenotypes.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号